Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Mar 19.
Published in final edited form as: Prostaglandins Other Lipid Mediat. 2016 Sep 17;132:31–40. doi: 10.1016/j.prostaglandins.2016.09.002

FUNCTIONAL SCREENING FOR G PROTEIN-COUPLED RECEPTOR TARGETS OF 14,15-EPOXYEICOSATRIENOIC ACID

Xuehong Liu 4, Zu-yuan Qian 4, Fuchun Xie 2, Wei Fan 4, Jonathan W Nelson 4, Xiangshu Xiao 2,4, Sanjiv Kaul 4, Anthony P Barnes 3,4, Nabil J Alkayed 1,2,4
PMCID: PMC6424572  NIHMSID: NIHMS940521  PMID: 27649858

Abstract

Epoxyeicosatrienoic acids (EETs) are potent vasodilators that play important roles in cardiovascular physiology and disease, yet the molecular mechanisms underlying the biological actions of EETs are not fully understood. Multiple lines of evidence suggest that the actions of EETs are in part mediated via G protein-coupled receptor (GPCR) signaling, but the identity of such a receptor has remained elusive. We sought to identify 14,15-EET-responsive GPCRs. A set of 105 clones were expressed in Xenopus oocyte and screened for their ability to activate cAMP-dependent chloride current. Several receptors responded to micromolar concentrations of 14,15-EET, with the top five being prostaglandin receptor subtypes (PTGER2, PTGER4, PTGFR, PTGDR, PTGER3-IV). Overall, our results indicate that multiple low-affinity 14,15-EET GPCRs are capable of increasing cAMP levels following 14,15-EET stimulation, highlighting the potential for cross-talk between prostanoid and other ecosanoid GPCRs. Our data also indicate that none of the 105 GPCRs screened met our criteria for a high-affinity receptor for 14,15-EET.

Keywords: Epoxyeicosatrienoic acid, EET, GPCR, CFTR, Prostaglandin E2, vasodilation

INTRODUCTION

Epoxyeicosatrienoic acids (EETs) are endogenous vasodilators in multiple vascular beds [1,2]. Potency of 14,15-EET as a vasodilator varies depending on the tissue and vessel size. For example, in bovine coronary arteries and rat mesenteric arteries, 14,15-EET has been shown to induce vasodilation in micromolar range [28] while high potency (low picomolar) vasodilation by EETs has been observed in canine and porcine coronary microvessels [1,9]. We have previously demonstrated that 14,15-EET is abundant in brain tissue, playing critical roles in both normal function and disease [10].

Multiple ligand binding studies suggest the existence of a high affinity (low nanomolar) receptor for 14-15-EET [8,1114]. To identify this putative high affinity receptor, we selected 150 candidate GPCRs based on their similarities to other lipid-sensing GPCRs. Only a subset of 105 receptors were successfully cloned and shown to display cell surface expression in Xenopus oocyte, which were subsequently screened for their ability to increase cAMP-dependent chloride current.

We focused on cAMP-based activity because it has been suggested that 14,15-EET exerts its vasodilator effect via Gs-coupled signaling leading to increased cAMP [8,12]. To monitor changes in cAMP in real time, we used the CFTR (Cystic Fibrosis Transmembrane Conductance Regulator) channel as a functional reporter of increased cAMP and activation of Protein Kinase A (PKA), as opening of CFTR channel requires phosphorylation by PKA [15,16]. In this case, the CFTR channel was co-expressed in an oocyte expression system along with each GPCR of interest. The utility of this method in studying GPCR signaling has been demonstrated previously [1719]. We also employed ERK activation assay and a β-arrestin recruitment assay as alternative methods to detect GPCR activation by 14,15-EET. Our results indicate that while none of the candidate receptors tested met our criteria for a high affinity Gs-coupled receptor for 14,15-EET, we were able to identify several previously unknown low affinity 14,15-EET receptors.

MATERIALS AND METHODS

Mouse mesenteric artery diameter measurement

Mouse mesenteric arteries were isolated, cut into 1–2 mm in length and mounted on a single vessel chamber (CH-1 Living Systems Instrumentation, Burlington, VT), secured between two glass micropipettes and tied with two knots on each end. Vessels were maintained at a constant pressure (80 mmHg) using a pressure servo system (Living Systems Instrumentation, Burlington, VT), and superfused continuously with MOPS buffered solution (in mM: 144 NaCl, 3.0 KCl, 2.5 CaCl2, 1.5 MgSO4, 2.0 MOPS, 5.0 glucose, 2.0 Pyruvate, 0.02 EDTA, 1.2 NaH2PO4) with a flow rate of 2.5 mL/min. The temperature of the perfusion chamber was maintained at 37°C using both stationary (TC-095, Living Systems Instrumentation, Burlington, VT) and inline heater (TC-344B, Warner Instruments, Hamden, CT). Vessel diameter was continuously monitored using a video dimension analyzer (Living Systems Instrumentation, Burlington, VT), digitized and recorded using AxonScope data acquisition software (Molecular Devices, Sunnyvale, CA)

HEK293 cell culture and transfection

Human Embryonic Kidney-293 cells (HEK 293; American Type Culture Collection, Manassas, VA) were grown in Dulbecco’s modified Eagle’s medium (DMEM, Invitrogen) supplemented with 10% fetal bovine serum (FBS). Cells were thawed, maintained in 5% CO2 at 37°C and used through passage 12. Cells grown in 12-well plates were transfected with 2.5 μg GPCR plasmids and Lipofectamine 2000 reagent (Invitrogen) at 50–70% confluency in media lacking antibiotics. Cells transfected with transfection solution alone without the plasmid served as controls.

GPCR vector construction

GPCR clones were obtained from DNASU Plasmid Repository (The Biodesign Institute, Arizona State University. Tempe, AZ) and UMR cDNA Resource Center (University of Missouri-Rolla, Rolla, MO). They were subcloned into a modified pcDNA3.1 vector containing both CMV and T7 promotors to permit both mammalian expression and in vitro transcription of each candidate receptor for oocyte expression. The expression vector also contains an in-frame hemagglutinin (HA) epitope tag that resulted in amino terminus tagging of each candidate receptor.

cAMP detection assay

cAMP concentrations of oocytes were detected using the Cyclic AMP XP® Assay Kit from Cell Signaling Technology (Danvers, MA) based on the manufacture’s protocol with a slight modification where 50 μM IBMX [18] was included in the stimulatory cocktail and 100 mM HCl [20] was included in the lysis buffer to minimize PDE activity in oocytes.

Oocyte expression system

The cRNAs for human CFTR (hCFTR) and candidate GPCRs were generated using Ambion mMessage mMachine T7 Ultra transcription kit (Ambion, Foster City, CA). Follicular membranes of Xenopus laevis oocytes were removed using 0.2 Wünsch units/mL Liberase TL (Roche Molecular Biochemicals, Indianapolis, IN) for 2 h in a Ca2+-free solution containing (mM): 82.5 NaCl, 2 KCl, 1 MgCl2, 5 HEPES, pH 7.5. Oocytes were sorted and maintained in a modified Barth’s solution containing (mM): 88 NaCl, 1 KCl, 0.82 MgSO4, 0.33 Ca(NO3)2 0.41 CaCl2, 2.4 NaHCO3, 5 HEPES, 5 HEPES-Na, and 250 mg/L Amikacin plus 150 mg/L Gentamicin at pH 7.5. Stage V to VI oocytes were injected with 50 nL cRNA mix containing CFTR RNA and GPCR RNA.

Whole-cell recordings were carried out using Oocyte 725 amplifier (Warner Instruments, Hamden, CT) and pClamp 8 software (Molecular Devices, Sunnyvale, CA). Individual oocytes were placed in a modified recording chamber (RC-1Z, Warner Instruments, Hamden, CT) and continuously superperfused with standard Frog Ringer’s (FR) solution (in mM: 98 NaCl, 2 KCl, 1 MgCl2, 1.8 CaCl2, 2.5 HEPES, 2.5 HEPES-Na) where they were maintained in the open circuit condition and the membrane potential was periodically ramped from −120 to +60 mV over 1.8 sec to obtain current-voltage relationships.

Detection of cell surface expression of GPCRs

Oocytes or HEK cells expressing candidate GPCRs were incubated for 1 h at 4°C in the dark in Frog Ringer’s solution and PBS containing 1 μg/mL Alexa Fluor 488 conjugated anti-HA antibody (ThermoFisher®, Grand Island, NY), respectively. Cells were washed three times prior to imagining using laser scanning confocal microscopy. Fig. 1 contains examples of surface labeling in oocytes. Oocytes expressing hCFTR that lacks the HA-tag was used as a control. We confirmed surface expression of 105 candidate receptors in oocytes (Table 1).

Figure 1.

Figure 1

Examples of surface detection of prostaglandin receptors expressed in oocytes. Receptors were labeled with a fluorophore (Alexa Fluor® 488 dye) and detected by confocal microscopy. Gray scale was used for clarity. All receptors showed above-background labeling when oocytes were injected with equal or greater amount of RNA than that used for electrophysiological experiments described below.

Table 1.

GPCRs with Confirmed Expression in Oocytes

ADRB2 CNR2 CCKBR LPAR3
CXCR4 CNR1 CCR1 B1AR
CYSLTR1 GPR68 (oGPR1) CCR3 Cmklr1
PTGER2 LPAR4 CHRM2 Mrgprf
PTGER4 GPR35 CHRM3 Galr2
GPCR132 PTGFR CHRM4 Opn3
PAR2 PTGDR GPCR4 Kiss1r
LTB4R PTGER3 HRH2 Gpr135
EDG4 (LPAR2) PTGER1 HTR1A Ccr10
P2RY2 GPCR17 HTR4 Gpr19
PTAFR GPCR18 HTR6 Lgr4
EDG3 GPCR20 LTB4R2 Sucnr1
EDG6 GPCR63 NPFFR1 Mrgpre
TBXA2R GPCR161 OPRL1 Gpr162
GPR31 P2RY1 GPCR21 Gpr107
GPR3 P2RY4 GPCR52 Gpr146
GPR61 P2RY6 GPCR141 Gpr137B
GPR85 SSTR2 GPCR148 Tpra1
GPR173 SSTR3 GPCR176 Gpr39
PGIR SSTR4 GPCR34 Npy1r
LPAR2 ADORA2 GPCR81 Adora2b
LPAR1 ADRB3 GPCR116 GPR32
S1P3 AGTR1 GPCR125 C5L2/C5AR2
S1P1 APLNR GPCR26 GPR101
S1P5 AVPR1A GPCR62 FPR3
S1P2 C5AR1 GPCR78 BONZO/CXCR6
ADORA1

ERK phosphorylation (ERKp) assay

HEK cells transfected with vector alone or with candidate receptors were serum starved for 1 hour prior to exposure to either vehicle (DMSO) or 1 μM 14,15-EET for 15 min. Cells were then lysed in lysis buffer containing 50 mM Tris (pH 7.4), 150 mM NaCl, 1% NP-40, 0.25% Na-deoxycholate, 1 mM ethylenediaminetetraacetic acid and protease inhibitor (Roche, Nutley, NJ). HEK proteins in SDS sample buffer (2% SDS, 10% glycerol, 80 mM Tris, pH 6.8,0.15 M β-mercaptoethanol, 0.02% bromphenol blue) were separated on 4–12% SDS–polyacrylamide gels (20 μg/lane) and transferred to polyvinylidene difluoride membranes. The membranes were blocked with 5% nonfat dry milk in TBST (10 mM Tris, pH 7.5,150 mM NaCl, 0.05% Tween 20) for 30 minutes at room temperature and incubated overnight at 4°C with 1:1000 rabbit anti-ERK (Cell Signaling, Danvers, MA) primary antibodies in dry milk.

The antigens were detected by the luminescence method (ECL-plus Western bloting detection kit) with peroxidase-linked anti-rabbit antibodies (GE Amersham, Lafayette, CO). The intensity of immunoblot bands was detected and quantified using the Fluor Chem FC2 Image Analysis System (Alpha Innotech, Wallingford, CT). Levels of total ERK and phosphorylated ERK were normalized using levels of beta-actin.

PathHunter β-arrestin enzyme fragment complementation (EFC) assay

The high throughput β-arrestin screening assay developed by DiscoveRx Corporation (Fremont, CA) makes use of two inactive complementary fragments of β-galactosidase (β-Gal) that are fused to β-arrestin and target GPCR, respectively. Receptor activation restores β-Gal activity when the two complementary β-Gal fragments merge as a result of formation of β-arrestin-G-protein complex. The restored β-Gal activity was measured by chemiluminescent PathHunter® Detection Reagents following stimulation by 1 μM 14,15-EET. A total of 241 GPCRs, including 73 orphan GPCRs were screened (https://www.discoverx.com/services/drug-discovery-evelopment-services/gpcrscan-gpcr-profiling).

Standard procedures employed by DiscoveRx Corporation (Fremont, CA) are summarized as follows: Cell Handling. PathHunter cell lines expressing each receptor were expanded from freezer stocks were seeded in a total volume of 20 μL into white walled, 384-well microplates and incubated at 37°C for 90 min prior to testing. Agonist Activity Assay. 5 μL of 5× 14,15-EET diluted in assay buffer was added to cells and incubated at 37°C for 90 min. Final assay vehicle concentration was 1% for all experiments. AntagonistActivity Assay. 5 μL of 5× 14,15-EET stock diluted in assay buffer was added to cells and incubated at 37°C for 30 min. Then, 5 μL of 6× EC80 agonist diluted in assay buffer was added to the cells and incubated at 37°C for 90 min. Signal Detection. Assay signal was generated through a single addition of 50% v/v of PathHunter Detection reagent cocktail. Two data points were collected for each receptor per assay. After one hour incubation at room temperature, microplates were read with a PerkinElmer Envision™ instrument for chemiluminescent signal detection. Data Analysis. Compound activity was analyzed using CBIS data analysis suite (ChemInnovation, CA). The activity was reported as the mean of two replica. Agonist activity for orphan receptors was determined as percentage activity (%Act) calculated using Eq (1):

%Act=100%RLUEETRLUvehicleRLUvehicle (1)

Agonist activity for receptors with known ligands was determined as percentage activity (%Act) calculated using Eq (2):

%Act=100%RLUEETRLUvehicleRLUmaxRLUvehicle (2)

Antagonist activity was determined as percentage inhibition (%Inh) calculated using Eq (3):

%Inh=100%(1RLUEETRLUvehicleRLUEC80RLUvehicle) (3)

where RLUEET is the relative mean luminescent unit determined for 14,15-EET, RLUvehicle is the mean RLU for the vehicle control, RLUmax is the mean max RLU for control agonist and RLUEC80 is the mean RLU of control agonist EC80.

Strategy for oocyte-based receptor screening

To efficiently screen a large number of receptors using the oocyte assay, we first confirmed surface expression of the receptors, then co-expressed each candidate receptor with CFTR in Xenopus oocytes and assayed receptor activity using an electrophysiological method (two-electrode-voltage clamp, TEVC). Oocytes were first exposed to 10 μM (forskolin. The response to forskolin served as an internal control for the level of cAMP-induced CFTR conductance given that forskolin activates adenylate cyclase directly and its action is independent of receptor activation. Oocytes were then exposed to 1 μM 14,15-EET. If 1 μM 14,15-EET induced an increase in conductance, the same oocyte would then be exposed to lower concentrations 14,15-EET.

Reagents

Forskolin, IBMX (Isobutylmethylxanthine), 14(R),15(S)-EET, 14,15-EEZE and prostaglandin E2 (PGE2) were purchased from Cayman Chemical (Ann Arbor, MI); 14,15(±)-EET was synthesized in-house [21]. Although the majority of the experiments were performed using 14,15(±)-EET, 14(R),15(S)-EET was occasionally used, with no qualitative differences observed between the two compounds thus they were labeled throughout non-discriminately as 14,15-EET or EET; CF172 (CFTRinh-172, 4-[4-Oxo-2-thioxo-3-(3-trifluoromethyl-phenyl)-thiazolidin-5-ylidenemethyl]-benzoic acid) was kindly provided by Dr. Robert Bridges (Rosalind Franklin University, Chicago, IL) and The Cystic Fibrosis Foundation (CFF).

Statistical analysis

Results were analyzed using the Student’s t-test or two-way ANOVA when appropriate. Differences with p < 0.05 were considered statistically significant. Data are presented as means±SEM.

RESULTS

14,15-EET induces vasodilation in U46619-preconstricted mouse mesenteric arteries

The vasodilatory effect of EETs has been reported in vessels from multiple species, including bovine, rat, porcine and canine vessels [19]. Similarly, we find that 14,15-EET induces vasorelaxation with an EC50 of 510 nM and maximal dilation to 68% in isolated mouse mesenteric arteries preconstricted to 26–56% of their baseline diameter by 20–40 nM U46619, a thromboxane A2 receptor agonist [22] (Fig. 2).

Figure 2.

Figure 2

14,15-EET (EET)-induced vasodilation. Dose-response curve obtained from mouse mesenteric arteries pre-constricted with thromboxane A2 agonist U46619 (n=4–6).

14,15-EET increases conductance of oocytes co-expressing CFTR and prostaglandin receptors

To determine which GPCR mediates the increase in intracellular cAMP by 14,15-EET, we co-expressed candidate GPCRs with CFTR, and used the latter as a reporter for GPCR/Gs/cAMP activation. As shown in Fig. 3, we first confirmed that in oocytes expressing CFTR alone, 14,15-EET had no effect on conductance. Vehicle (ethanol) also had no effect on conductance of uninjected oocytes or those injected with CFTR alone or together with receptors (not shown).

Figure 3.

Figure 3

14,15-EET (EET) had no effect on conductance of oocytes expressing hCFTR alone. An oocyte expressing hCFTR is exposed to 10 μM forskolin (white bars and circles). After a wash, it was exposed to 10 nM, 100 nM and 1 μM EET (gray bars and circles), sequentially.

Thorough screening of each candidate receptor resulted in the identification of several EET-responsive receptors with the top five being prostaglandin receptor subtypes (PTGER4, PTGER2, PTGFR, PTGDR and PTGER3IV) (Fig. 4). Because conductance changes in response to 14,15-EET were only induced when these receptors were co-expressed with CFTR, we conclude that stimulation of CFTR conductance by 14,15-EET is solely due to the activation of the receptors. However, we also found that none of these receptors meet our criteria for a high affinity Gs-coupled receptor because micromolar concentrations are needed to achieve maximum stimulation.

Figure 4.

Figure 4

14,15-EET (EET) increased conductance in oocytes co-expressing hCFTR and (A) PTGER2, (B) PTGER4, (C) PTGFR, (D) PTGDR, or (E) PTGER3IV. In each case, oocytes co-expressing hCFTR and a candidate receptor were exposed to 10 μM forskolin (white bars and circles), 1 μM EET (dark gray bars and circles) and 100 nM 14,15-EET (gray bars and circles), sequentially with washes in-between. Normalized data with respect to forskolin-induced changes are summarized on the right side of each panel.

14,15-EET increases ERK phosphorylation in HEK293 cells expressing PTGER2 or PTGER4

To complement the results from the oocyte assay, we implemented another measure used extensively to study GPCR activation [23,24], namely ERK phosphorylation (pERK). As shown in Fig. 5, significantly affected levels of pERK were only seen in 14,15-EET treated cells transfected with either PTGER2 or PTGER4. The smaller effects of 14,15-EET on cells transfected with PTGDR, PTGFR and PTGER3IV might have been compromised due to reduced surface expression in HEK cells (Fig. 6). In this regard, it has been shown previously that oocytes are capable of expressing exogenous proteins that are poorly expressed in mammalian cells such as HEK cells [25].

Figure 5.

Figure 5

Summary of 14,15-EET-induced pERK in cultured HEK293 cells expressing prostaglandin receptor subtypes (n=3 each).

Figure 6.

Figure 6

Examples of surface detection of PGE2 receptors transfected in HEK cells. Receptors were labeled with an anti-HA antibody coupled to the fluorophore Alexa 488 and detected by confocal microscopy. Images were obtained under identical conditions. Scale bar in upper left image is 10 microns and applies to all images.

14,15-EET is a less efficacious agonist than PGE2 in stimulating CFTR conductance in oocytes co-expressing CFTR and prostaglandin receptors

Our finding that 14,15-EET activates several prostaglandin receptors raises the question of whether the effect of 14,15-EET is comparable to that induced by prostaglandin E2 (PGE2). We compared the effects of 1 μM 14,15-EET and 10 nM PGE2 in the same oocytes co-expressing CFTR and prostaglandin receptors. As shown in Fig. 7, we first confirmed that oocytes expressing CFTR alone had no conductance response following application of 10 nM PGE2. However, in oocytes co-expressing CFTR and the receptors, a 100 fold lower concentration of PGE2 was capable of eliciting a larger or similar increase in conductance compared to that elicited by 1 μM 14,15- EET (Fig. 8). Because PGE2 only induced changes in conductance when these receptors were co-expressed with CFTR, our results indicate that the stimulation of CFTR conductance by PGE2 was solely due to the activation of the receptors (the order of exposure to the reagents did not alter outcome, data not shown).

Figure 7.

Figure 7

PGE2 had no effect on conductance of oocytes expressing hCFTR alone. An oocyte expressing hCFTR was exposed to 10 μM forskolin (white bars and circles). After a wash, it was exposed to 10 nM PGE2.

Figure 8.

Figure 8

14,15-EET (EET) is less efficacious than PGE2 to increase conductance in oocytes co-expressing hCFTR and (A) PTGER2, (B) PTGER4, (C) PTGFR, (D) PTGDR or (E) PTGER3IV. In each case, oocytes co-expressing hCFTR and a candidate receptor were exposed to 10 μM forskolin (white bar and circles) followed by 1 μM and 100 nM EET (gray bar and circles) and 10 nM PGE2 (dark gray bar and circles) sequentially with washes in between. Normalized data in respect to forskolin-induced changes are summarized on the right side of each panel.

Effect of PGE2 on intracellular cAMP levels

Using forskolin stimulated increase in CFTR conductance as an internal control for the level of CFTR expression and activation due to elevation of intracellular cAMP level, we found the avergae ratio between the conductance induced by 10 nM PGE2 and that induced by 10 μM forskolin was 18±4 (n=4), 13±9 (n=3), 3.5±0.6 (n=4), 0.7±0.5 (n=4) and −0.4±0.1 (n=4) in oocytes co-expressing CFTR with PTGER2, PTGER4, PTGFR, PTGDR, or PTGER3IV, respectively. This result is consistent with idea that PGE2 is a preferred agonist that activates PTGER2 and PTGER4 and PTGDR via Gs-coupled receptors signaling via cAMP pathway [2629].

PTGER3 has multiple isoforms that couple to different signaling pathways [27,30]. A synthetic EP3 agonist M&B28767 has been shown to increase cAMP in COS-7 expressing PTGER3IV with a concentration greater than 1 μM and in CHO cells transfected with PTGER3IV with a concentration greater than 100 nM [30]. However, we are not aware of any study demonstrating the stimulatory effect of PGE2 on cAMP in cells transfected with either PTGER3IV or PTGFR. We therefore, used a commercial assay to determine if PGE2 increases levels of cAMP in oocytes expressing PTGER2, PTGFR, and PTGER3IV. As shown in Fig. 9, however, we did not observe a change in cAMP in oocytes expressing PTGFR and PTGER3IV. We were also unable to detect changes of cAMP in HEK cells transfected with PTGFR and PTGER3IV (data not shown). The only increase in cAMP by PGE2 we observed was in oocytes expressing PTGER2. Because electrophysiological recording in oocytes may have a higher sensitivity in detecting changes in oocyte intracellular cAMP (note an 18 fold increase detected by TEVC in oocytes coexpressing CFTR and PTGER2 vs. < 2 fold increase detected by the cAMP assay in oocytes expressing PTGER2), and to further investigate the role of cAMP in PGE2-mediated effects, we combined electrophysiological recordings with pharmacological inhibitors of cAMP pathway. Specifically, we compared the effects of PGE2 on the conductance of oocytes co-expressing CFTR and receptors of interest with and without pretreatment with a known PKA inhibitor, H89. Using a concentration previously shown to partially block PKA activity in oocytes [31], we found that pretreatment with 50 μM H89 eliminated more than 90% of PGE2-induced conductance in each paired experiment (Fig. 10).

Figure 9.

Figure 9

Effect of PGE2 on cAMP levels in oocytes expressing PTGER2, PTGFR and PTGER3IV. Oocytes were injected with 20 ng RNA and divided into experimental (black bars) and control groups (white bars). Three oocytes were pooled as one data point. On day 4 following injection, oocytes in experimental groups were treated with 10 nM PGE2 plus 50 μM IBMX for 15 min. cAMP levels in both groups were then assayed per manufacture’s protocol.

Figure 10.

Figure 10

Effect of PKA inhibitor H89 on conductance of oocytes co-expressing hCFTR and (A) PTGER2, (B) PTGFR, or (C) PTGER3IV. Oocytes were pretreated with 50 μM H89 for a minimum of 10 min. For each pair, control and H89-pretreated oocytes were exposed to 10 nM PGE2 (black bars) as soon as stable background conductance was established (white bars). 50 μM H89 was present at all times in the superfusate for H89-pretreated oocytes. P-values were less than 0.05 for panels A and C and it was 0.06 for panel B.

EEZE (14,15-EE-5(Z)-E) does not behave as a selective antagonist for 14,15-EET at the PTGER2 receptor

14,15-EEZE was initially identified as a selective antagonist for 14,15-EET [32]. In bovine coronary arteries and rat mesenteric artery, 14,15-EEZE blunted vasorelaxation induced by 14,15-EET [6,32]. However, it has also been shown that 14,15-EEZE can induce vasorelaxation [32,33]. Because PTGER2 has been suggested as a receptor for 14,15-EET [6], we carried out experiments to test the effect of 14,15-EEZE on PTGER2. We first determined that 14,15-EEZE had no effect on oocytes expressing CFTR alone (Fig. 11A). However, in oocytes co-expressing CFTR and PTGER2, 14,15-EEZE induced a robust increase in CFTR conductance, and pretreatment of 14,15-EEZE prevented additional stimulation by 14,15-EET in a concentration dependent manner (Fig. 11B&C), suggesting that 14,15-EEZE, like 14,15-EET, could be an agonist or partial agonist for PTGER2. To confirm that the increase in conductance was due to stimulation of CFTR channels, we also exposed oocytes to 10 μM CF172 [34], a specific inhibitor for CFTR channel. As shown towards the end of the experiments, CF172 induced a rapid decrease in conductance, demonstrating that activation of PTGER2 by 14,15-EEZE was solely due to the stimulation of CFTR channels.

Figure 11.

Figure 11

14,15-EEZE (EEZE) is an agonist/partial agonist for PTGER2. (A) An oocyte expressing hCFTR was exposed to 10 μM forskolin (white bar and circles), vehicle (white bar and triangles, 1μM and 10 μM EEZE. Summary of normalized data in respect to forskolin-induced changes are shown at right. (B) An oocyte co-expressing hCFTR and PTGER2 was exposed to 1 μM EET (gray bar and circles), 1 μM EEZE (dark gray bar and circles, and 1 μM EET in the presence of 1μM EEZE. Followed by exposure to 10 μM CF172. (C) A similar experiment carried out in an oocyte co-expressing hCFTR and PTGER2 as shown in panel B, except 10 μM EEZE was used in place of 1 μM EEZE. Summary of normalized data in respect to EET-induced changes for panel B&C are shown on the right for each panel.

The stimulatory effect of 14,15-EET and 14,15-EEZE are not due to PGE2 synthesis

An alternative explanation to direct binding and activation of the prostaglandin receptors by 14,15-EET is that it instead exerts its effect by stimulating PGE2 synthesis in oocytes. We therefore investigated the effect of the prostacyclin inhibitor indomethacin on PGE2-stimulated CFTR conductance. As shown in Fig. 12, pre-exposure of oocytes co-expressing CFTR and PTGER2 to indomethacin had no significant effect on the changes in conductance induced by 14,15-EET (Fig. 12A) or 14,15-EEZE (Fig. 12B), suggesting that PGE2 synthesis is unlikely to mediate the stimulatory effect of 14,15-EET or 14,15-EEZE on PTGER2.

Figure 12.

Figure 12

Effects of 14,15-EET (EET) and 14,15-EEZE (EEZE) were not due to PGE2 synthesis. (A) An oocyte co-expressing hCFTR and PTGER2 was exposed to 1 μM EET (gray bar and circles), 10 μM indomethacin (IMC) (dark gray bar and circles, and 1 μM EET in the presence of 10 μM IMC, followed by exposure to 10 μM CF172. (B) A similar experiment carried out in an oocyte co-expressing hCFTR and PTGER2 as shown in panel B, except 10 μM EEZE was used in place of 1 μM EET.

Putative GPCR screening based on β-Arrestin recruitment

To expand our search for a putative receptor for 14,15-EET, we took advantage of a high- throughput β-arrestin screening assay using 1 μM 14,15-EET as a probe. The DiscoveRx receptor library contained a total of 241 candidate receptors, including 174 candidate receptors not screened in the oocyte assay. All receptors were screened for the agonist activity of 14,15-EET. Those with known ligands were also screened for the antagonist activity of 14,15-EET. Based on the mean values of two replicas, none of the receptors exhibited more than 30% increase in activity following stimulation by 1 μM 14,15-EET in the agonist assay. Receptors exhibiting an increase in activity for at least 15% are shown in Table 2. Similarly, none of the receptors exhibited more than 50% decrease in activity when 1 μM 14,15-EET was employed as an antagonist and those receptors displaying more than 28% inhibition by 1 μM 14,15-EET are indicated in Table 3.

Table 2.

Top Ranked Receptors Based on Stimulation of β-Arrestin Recruitment by 14,15-EET

Receptor %activity
CCKBR 26%
CNR2 20%
CXCR4 17%
GHSR1B 19%
GPR61 15%
GPR139 15%
HTR1B 15%
P2RY8 15%

Table 3.

Top Ranked Receptors based on Inhibition by 14,15-EET

Receptor %inhibition
FSHR −47%
GPR109A −41%
BRS3 −32%
PROKR2 −32%
HRH4 −30%
CMKLR1 −28%
CALCR-RAMP3 −28%
AVPR1B −28%
PRLHR −28%

14,15-EET increases ERK phosphorylation in HEK293 cells expressing CXCR4 or CMKLR1

A subset of receptors exhibiting responsiveness to 14,15-EET in the DiscoveRx screen were found not to be responsive to 14,15-EET in the oocyte system (marked in Italic Bold in Tables 2 and 3). Therefore, we further examined these candidates using the ERK phosphorylation assay. As shown in Fig. 13, 1 μM 14,15-EET significantly increased pERK in HEK293 cells expressing CXCR4 or CMKLR1.

Figure 13.

Figure 13

Summary of 14,15-EET-induced pERK in cultured HEK293 cells expressing receptors that exhibited highest activity in β-arrestin assay (n=3 each).

DISCUSSION

Identification of low affinity receptors for 14,15-EET

Greater insight into the molecular mechanisms underlying EETs’ vasodilatory actions are critical to understanding their roles in vascular physiology and disease. Several studies have shown that 14,15-EET can dilate isolated vessels within the micromolar range [28]. One study found that the vasodilatory effect of 14,15-EET in rat mesenteric arteries was mediated through PTGER2 via Gs-coupled signaling [6].

Our current findings support the idea that PTGER2 is involved in mediating the micromolar effects of 14,15-EET [6]. It is not clear why these investigators did not observe a role for PTGER4 in mediating the vasodilatory effect of 14,15-EET, although their Western blots showed similar abundance for PTGER2 and PTGER4 in primary cultured rat mesenteric artery smooth muscle cells. Both PTGER2 and PTGER4 are known to couple to Gαs, causing an increase in intracellular cAMP in response to agonist [28,35].

Our observation that PGE2 treatment of oocytes co-expressing CFTR and PTGDR results in a stimulatory effect on CFTR conductance is consistent with the notion that PTGDR is a Gs-coupled receptor [29,36]. PGE2 has been shown to stimulate cAMP production in HEK cells stably expressing human PTGDR with an EC50 of 84 nM [29]. However, in another study, it was showed that EC50 was greater than 500 nM for PGE2 in stimulating cAMP in HEK293 cells stably expressing human PTGDR [36]. It is not obvious why the difference in EC50 varied by nearly tenfold between the two studies. It is possible that difference in the sources for the cells and detection reagents used may attribute to such difference.

PTGER3 is known to have multiple subtypes that couple to different signaling pathways [28,30]. Our current observation that PGE2 stimulation of PTGER3IV increased CFTR conductance in oocytes co-expressing CFTR and PTGER3IV suggests that PGE2 activation of PTGER3IV may increase intracellular cAMP. Inhibition of PGE2 effect by PKA inhibitor H89 is also consistent with such hypothesis. Although H89 is known to inhibit other kinases in some cell types [3739], we are not aware of any study linking these kinases to the activation of CFTR channels.

It is unclear why we were not able to detect a change in cAMP due to PTGER3IV activation using an conventional cAMP assay. It is possible that the sensitivity of our electrophysiological approach (TEVC), detects small changes that were not detected by conventional ELISA assays. On the other hand, although the change in CFTR conductance due to PGE2 activation of PTGER3IV were significantly different from the controls, the extend of the change due to activation of PTGER3IV is much smaller than that due to activation of PTGER2 as demonstrated when their effects were compared with that induced by forskolin. Therefore, even if activation of PTGER3IV increases intracellular cAMP, the physiological significance of such effect can not be evaluated without additional studies.

Our finding that activation of PTGFR stimulated CFTR conductance raises an interesting question regarding the signaling pathways associated with PTGFR, which is best known as a Gq-coupled receptor that increases intracellular Ca2+ when activated by its specific ligand, PGF [40]. We are not aware of any previous study specifically examining the effect of activation of PTGFR by PGE2 on intracellular cAMP concentration. As an agonist, PGE2 is not as potent as PGF for PTGFR based on a competitive binding assay against [3H]PGF (Kd=1 nM) [40]. Using membranes from COS cells expressing PTGFR, these authors found that the IC50 for PGE2 was 85 nM. In oocytes expressing PTGFR, they observed a dose-dependent increase in intracellular Ca2+ stimulated by PGE2, although PGE2 did not induce any detectable change in intracellular Ca2+ at 10 nM, unlike PGF [40].

The increase in CFTR conductance due to activation of PTGFR by PGE2 was significant as detected by monitoring using TEVC in real time. The inhibitory effect of PKA inhibitor H89 on PGE2 stimulated CFTR conductance is also consistent with the idea that activation of PTGFR may increase cAMP. However, the extent of the increase in CFTR conductance due to activation of PTGFR is much smaller than that due to activation of PTGER2 as demonstrated when their effects were normalized to forskolin-induced conductance. Furthermore, since we were not able to detect a change in cAMP directly using a conventional cAMP assay, additional studies using different cells type and different techniques may be necessary to validate whether PTGFR can couple to cAMP dependent signaling pathway.

Overall, our current study has raised interesting questions regarding the interaction between 14,15-EET and prostaglandin receptors. Given the relatively low plasma concentration of EETs [41,42], the physiological relevance of the micromolar dilatory effect of EETs remains unclear [1].

An argument for the existence of a high affinity receptor for 14,15-EET

A high affinity receptor for 14-15-EET was implicated in membrane binding studies (low nanomolar) [3,8,1114]. High potency EET-dependent vasodilation has been reported in canine and porcine coronary microvessels [1,9]. We found that 14,15-EET induced vasorelaxation in mouse mesenteric arteries with an EC50 of 510 nM. Although EET-responsive receptors discovered in the current study are all low affinity, our results do not necessarily exclude the possibility of the existence of a high affinity receptor for 14,15-EET.

EEZE as a Partial Agonist

Although 14,15-EEZE has been suggested initially as a selective antagonist for EETs [32], additional studies have also shown that 14,15-EEZE can act as a partial agonist for endothelial derived hyperpolarizing factor (EDHF), exerting vasodilatory effect on Bovine coronary artery [32] or mouse mesenteric arteries [33]. In agreement with these studies, we found that 14,15-EEZE has a stimulatory effect on the conductance of oocytes co-expressing CFTR and PTGER2. Our observation is consistent with the notion that 14,15-EEZE is an agonist or partial agonist for PTGER2.

PGE2 synthesis is not responsible for the stimulatory effects of 14,15-EET and 14,15-EEZE

Because PGE2 is a naturally occurring physiological molecule, it is possible that 14,15-EET and 14,15-EEZE treatment could stimulate PGE2 production in oocytes rather than directly activate the receptors. However, our results demonstrate that 14,15-EET and 14,15-EEZE directly activate the receptor because their stimulatory effects were unaffected by pre-treatment with the PGE2 synthesis inhibitor indomethacin [43]. In addition, it has been shown that 14,15-EEZE paritially blocked the dilatory effect of PGE2 in rat mesenteric arteries [6]. Furthermore, the time course of the effects of 14,15-EET and 14,15-EEZE observed in oocytes was much more rapid than PGE2 synthesis through the COX enzyme that has a half time in the scale of hours [44]

Importance of assay selection in receptor screening

We employed three different assays in the current study to screen for a putative receptor for 14,15-EET. Among the five low affinity EET-responsive receptors we identified using the oocyte expression assay, only two were significantly stimulated by 14,15-EET in the pERK assay. In addition, among the five receptors tested using both β-arrestin and pERK assays, only two were significantly stimulated by 14,15-EET in the pERK assay. There are at least three possible explanations for the discrepancies in results obtained by different assays. First, oocytes tend to be a more robust system to express some exogenous proteins than mammalian cells [25] and it has very low background noise, which makes it a very sensitive assay. Oocytes assay has the additional advantage of allowing real time studies under physiological conditions as demonstrated in the current study. Second, different methods are likely to have different detection limit for monitoring receptor activation. Third, the three assays used in the current study were performed in different cell types, which may utilize different G-protein coupling and signal transduction pathways. In our view, in cases when the coupling mechanisms are unknown, it is crucial to employ multiple assays for receptor screening.

CONCLUSION

Our current study confirms the existence of several low affinity receptors for 14,15-EET. None of the receptors screened in our study met the criteria for a high affinity Gs-coupled receptor for 14,15-EET. However, our results do not rule out the existence of a putative high affinity receptor for 14,15-EET. Our results also do not rule out the potential involvement of other signaling pathways in mediating the effect of 14,15-EET.

Acknowledgments

FUNDING

This work is supported by the Knight Cardiovascular Institute at Oregon &Health & Science University, Portland, OR.

ABBREVIATIONS

14,15-EET (EET)

14,15-Epoxyeicosatrienoic acids

14,15-EEZE

14,15-Epoxyeicosa-5(Z)-enoic Acid

VSMCs

vascular smooth muscle cells

GPCR

G protein-coupled receptor

CFTR

Cystic Fibrosis Transmembrane Conductance Regulator

PKA

Protein Kinase A

PTG

prostaglandin

PGE2

prostaglandin E2

IMC

indomethacin

CF172

CFTRinh-172 (5-[(4-Carboxyphenyl)methylene]-2-thioxo-3-[(3-trifluoromethyl)phenyl-4-thiazolidinone)

IBMX

3-isobutyl-1-methylxanthine

TEVC

two- electrode-voltage clamp

Footnotes

CONFLICT OF INTEREST

The authors declare that there are no conflicts of interest.

References

  • 1.Oltman CL, Weintraub NL, VanRollins M, Dellsperger KC. Epoxyeicosatrienoic acids and dihydroxyeicosatrienoic acids are potent vasodilators in the canine coronary microcirculation. Circulation research. 1998;83:932–939. doi: 10.1161/01.res.83.9.932. [DOI] [PubMed] [Google Scholar]
  • 2.Yang C, Yang J, Xu X, Yan S, Pan S, Pan X, Zhang C, Leung GP. Vasodilatory effect of 14,15-epoxyeicosatrienoic acid on mesenteric arteries in hypertensive and aged rats. Prostaglandins & other lipid mediators. 2014;112:1–8. doi: 10.1016/j.prostaglandins.2014.05.001. [DOI] [PubMed] [Google Scholar]
  • 3.Chen Y, Falck JR, Manthati VL, Jat JL, Campbell WB. 20-Iodo-14,15-epoxyeicosa-8(Z)-enoyl-3-azidophenylsulfonamide: photoaffinity labeling of a 14,15-epoxyeicosatrienoic acid receptor. Biochemistry. 2011;50:3840–3848. doi: 10.1021/bi102070w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Falck JR, Krishna UM, Reddy YK, Kumar PS, Reddy KM, Hittner SB, Deeter C, Sharma KK, Gauthier KM, Campbell WB. Comparison of vasodilatory properties of 14,15-EET analogs: structural requirements for dilation. American journal of physiology Heart and circulatory physiology. 2003;284:H337–349. doi: 10.1152/ajpheart.00831.2001. [DOI] [PubMed] [Google Scholar]
  • 5.Gauthier KM, Falck JR, Reddy LM, Campbell WB. 14,15-EET analogs: characterization of structural requirements for agonist and antagonist activity in bovine coronary arteries. Pharmacological research. 2004;49:515–524. doi: 10.1016/j.phrs.2003.09.014. [DOI] [PubMed] [Google Scholar]
  • 6.Yang C, Kwan YW, Au AL, Poon CC, Zhang Q, Chan SW, Lee SM, Leung GP. 14,15-Epoxyeicosatrienoic acid induces vasorelaxation through the prostaglandin EP(2) receptors in rat mesenteric artery. Prostaglandins & other lipid mediators. 2010;93:44–51. doi: 10.1016/j.prostaglandins.2010.06.004. [DOI] [PubMed] [Google Scholar]
  • 7.Yang C, Pan S, Yan S, Li Z, Yang J, Wang Y, Xiong Y. Inhibitory effect of 14,15-EET on endothelial senescence through activation of mTOR complex 2/Akt signaling pathways. Int J Biochem Cell Biol. 2014;50:93–100. doi: 10.1016/j.biocel.2014.02.020. [DOI] [PubMed] [Google Scholar]
  • 8.Yang W, Tuniki VR, Anjaiah S, Falck JR, Hillard CJ, Campbell WB. Characterization of epoxyeicosatrienoic acid binding site in U937 membranes using a novel radiolabeled agonist, 20-125i-14,15-epoxyeicosa-8(Z)-enoic acid. J Pharmacol Exp Ther. 2008;324:1019–1027. doi: 10.1124/jpet.107.129577. [DOI] [PubMed] [Google Scholar]
  • 9.Zhang Y, Oltman CL, Lu T, Lee HC, Dellsperger KC, VanRollins M. EET homologs potently dilate coronary microvessels and activate BK(Ca) channels. American journal of physiology Heart and circulatory physiology. 2001;280:H2430–2440. doi: 10.1152/ajpheart.2001.280.6.H2430. [DOI] [PubMed] [Google Scholar]
  • 10.Iliff JJ, Jia J, Nelson J, Goyagi T, Klaus J, Alkayed NJ. Epoxyeicosanoid signaling in CNS function and disease. Prostaglandins & other lipid mediators. 2010;91:68–84. doi: 10.1016/j.prostaglandins.2009.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Chen Y, Falck JR, Tuniki VR, Campbell WB. 20-125Iodo-14,15-epoxyeicosa-5(Z)-enoic acid: a high-affinity radioligand used to characterize the epoxyeicosatrienoic acid antagonist binding site. J Pharmacol Exp Ther. 2009;331:1137–1145. doi: 10.1124/jpet.109.157818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wong PY, Lai PS, Falck JR. Mechanism and signal transduction of 14 (R), 15 (S)-epoxyeicosatrienoic acid (14,15-EET) binding in guinea pig monocytes. Prostaglandins & other lipid mediators. 2000;62:321–333. doi: 10.1016/s0090-6980(00)00079-4. [DOI] [PubMed] [Google Scholar]
  • 13.Wong PY, Lai PS, Shen SY, Belosludtsev YY, Falck JR. Post-receptor signal transduction and regulation of 14(R),15(S)-epoxyeicosatrienoic acid (14,15-EET) binding in U-937 cells. Journal of lipid mediators and cell signalling. 1997;16:155–169. doi: 10.1016/s0929-7855(97)00005-9. [DOI] [PubMed] [Google Scholar]
  • 14.Wong PY, Lin KT, Yan YT, Ahern D, Iles J, Shen SY, Bhatt RK, Falck JR. 14(R),15(S)-epoxyeicosatrienoic acid (14(R),15(S)-EET) receptor in guinea pig mononuclear cell membranes. Journal of lipid mediators. 1993;6:199–208. [PubMed] [Google Scholar]
  • 15.Wilkinson DJ, Strong TV, Collins FS, Dawson DC. CFTR activation: Stimulatory and inhibitory phosphorylation sites in the R domain. Pediatric Pulmonology. 1994;(Supplement 10):179–180. doi: 10.1152/ajplung.1997.273.1.L127. doi. [DOI] [PubMed] [Google Scholar]
  • 16.Gadsby DC, Nairn AC. Control of CFTR channel gating by phosphorylation and nucleotide hydrolysis. Physiol Rev. 1999;79:S77–S107. doi: 10.1152/physrev.1999.79.1.S77. [DOI] [PubMed] [Google Scholar]
  • 17.Grygorczyk R, Abramovitz M, Boie Y, Bastien L, Adam M. Detection of adenylate cyclase-coupled receptors in Xenopus oocytes by coexpression with cystic fibrosis transmembrane conductance regulator. Anal Biochem. 1995;227:27–31. doi: 10.1006/abio.1995.1248. [DOI] [PubMed] [Google Scholar]
  • 18.Liu X, Grandy DK, Janowsky A. Ractopamine, a livestock feed additive, is a full agonist at trace amine-associated receptor 1. J Pharmacol Exp Ther. 2014;350:124–129. doi: 10.1124/jpet.114.213116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Uezono Y, Bradley J, Min C, McCarty NA, Quick M, Riordan JR, Chavkin C, Zinn K, Lester HA, Davidson N. Receptors that couple to 2 classes of G proteins increase cAMP and activate CFTR expressed in Xenopus oocytes. Receptors Channels. 1993;1:233–241. [PubMed] [Google Scholar]
  • 20.Gill A, Hammes SR. G beta gamma signaling reduces intracellular cAMP to promote meiotic progression in mouse oocytes. Steroids. 2007;72:117–123. doi: 10.1016/j.steroids.2006.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Xie F, Li BX, Alkayed NJ, Xiao X. Synthesis of 14,15-EET from Arachidonic Acid Using Urea-Hydrogen Peroxide as the Oxidant. Synthetic Communications. 2015;45:105–110. doi: 10.1080/00397911.2014.956369. [DOI] [Google Scholar]
  • 22.Abramovitz M, Adam M, Boie Y, Carriere M, Denis D, Godbout C, Lamontagne S, Rochette C, Sawyer N, Tremblay NM, Belley M, Gallant M, Dufresne C, Gareau Y, Ruel R, Juteau H, Labelle M, Ouimet N, Metters KM. The utilization of recombinant prostanoid receptors to determine the affinities and selectivities of prostaglandins and related analogs. Biochim Biophys Acta. 2000;1483:285–293. doi: 10.1016/s1388-1981(99)00164-x. doi. [DOI] [PubMed] [Google Scholar]
  • 23.Belcheva MM, Coscia CJ. Diversity of G protein-coupled receptor signaling pathways to ERK/MAP kinase. Neuro-Signals. 2002;11:34–44. doi: 10.1159/000057320. doi:57320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Garbison KE, Heinz BA, Lajiness ME, Weidner JR, Sittampalam GS. In: Assay Guidance Manual. Sittampalam GS, et al., editors. Eli Lilly & Company and the National Center for Advancing Translational Sciences; 2004. [PubMed] [Google Scholar]
  • 25.Denning GM, Anderson MP, Amara JF, Marshall J, Smith AE, Welsh MJ. Processing of mutant cystic fibrosis transmembrane conductance regulator is temperature-sensitive [see comments] Nature. 1992;358:761–764. doi: 10.1038/358761a0. [DOI] [PubMed] [Google Scholar]
  • 26.Bastepe M, Ashby B. The long cytoplasmic carboxyl terminus of the prostaglandin E2 receptor EP4 subtype is essential for agonist-induced desensitization. Mol Pharmacol. 1997;51:343–349. doi: 10.1124/mol.51.2.343. [DOI] [PubMed] [Google Scholar]
  • 27.Bastien L, Sawyer N, Grygorczyk R, Metters KM, Adam M. Cloning, functional expression, and characterization of the human prostaglandin E2 receptor EP2 subtype. J Biol Chem. 1994;269:11873–11877. [PubMed] [Google Scholar]
  • 28.Sugimoto Y, Narumiya S. Prostaglandin E receptors. J Biol Chem. 2007;282:11613–11617. doi: 10.1074/jbc.R600038200. [DOI] [PubMed] [Google Scholar]
  • 29.Wright DH, Metters KM, Abramovitz M, Ford-Hutchinson AW. Characterization of the recombinant human prostanoid DP receptor and identification of L-644,698, a novel selective DP agonist. Br J Pharmacol. 1998;123:1317–1324. doi: 10.1038/sj.bjp.0701708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kotani M, Tanaka I, Ogawa Y, Usui T, Mori K, Ichikawa A, Narumiya S, Yoshimi T, Nakao K. Molecular cloning and expression of multiple isoforms of human prostaglandin E receptor EP3 subtype generated by alternative messenger RNA splicing: multiple second messenger systems and tissue-specific distributions. Mol Pharmacol. 1995;48:869–879. [PubMed] [Google Scholar]
  • 31.Stahl K, Stahl M, de Jonge HR, Forrest JN., Jr ANP and CNP activate CFTR expressed in Xenopus laevis oocytes by direct activation of PKA. Journal of receptor and signal transduction research. 2015;35:493–504. doi: 10.3109/10799893.2015.1015738. [DOI] [PubMed] [Google Scholar]
  • 32.Gauthier KM, Deeter C, Krishna UM, Reddy YK, Bondlela M, Falck JR, Campbell WB. 14,15-Epoxyeicosa-5(Z)-enoic acid: a selective epoxyeicosatrienoic acid antagonist that inhibits endothelium-dependent hyperpolarization and relaxation in coronary arteries. Circulation research. 2002;90:1028–1036. doi: 10.1161/01.res.0000018162.87285.f8. [DOI] [PubMed] [Google Scholar]
  • 33.Harrington LS, Falck JR, Mitchell JA. Not so EEZE: the ‘EDHF’ antagonist 14, 15 epoxyeicosa-5(Z)-enoic acid has vasodilator properties in mesenteric arteries. Eur J Pharmacol. 2004;506:165–168. doi: 10.1016/j.ejphar.2004.09.062. [DOI] [PubMed] [Google Scholar]
  • 34.Ma T, Thiagarajah JR, Yang H, Sonawane ND, Folli C, Galietta LJ, Verkman AS. Thiazolidinone CFTR inhibitor identified by high-throughput screening blocks cholera toxin-induced intestinal fluid secretion. J Clin Invest. 2002;110:1651–1658. doi: 10.1172/JCI16112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hatae N. Cooperation of two subtypes of PGE2 receptor, Gi coupled EP3 and Gs coupled EP2 or EP4 subtype. Yakugaku zasshi : Journal of the Pharmaceutical Society of Japan. 2003;123:837–843. doi: 10.1248/yakushi.123.837. [DOI] [PubMed] [Google Scholar]
  • 36.Boie Y, Sawyer N, Slipetz DM, Metters KM, Abramovitz M. Molecular cloning and characterization of the human prostanoid DP receptor. J Biol Chem. 1995;270:18910–18916. doi: 10.1074/jbc.270.32.18910. doi. [DOI] [PubMed] [Google Scholar]
  • 37.Davies SP, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J. 2000;351:95–105. doi: 10.1042/0264-6021:3510095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lochner A, Moolman JA. The many faces of H89: a review. Cardiovascular drug reviews. 2006;24:261–274. doi: 10.1111/j.1527-3466.2006.00261.x. [DOI] [PubMed] [Google Scholar]
  • 39.Murray AJ. Pharmacological PKA inhibition: all may not be what it seems. Science signaling. 2008;1:re4. doi: 10.1126/scisignal.122re4. [DOI] [PubMed] [Google Scholar]
  • 40.Abramovitz M, Boie Y, Nguyen T, Rushmore TH, Bayne MA, Metters KM, Slipetz DM, Grygorczyk R. Cloning and expression of a cDNA for the human prostanoid FP receptor. J Biol Chem. 1994;269:2632–2636. [PubMed] [Google Scholar]
  • 41.Karara A, Wei S, Spady D, Swift L, Capdevila JH, Falck JR. Arachidonic acid epoxygenase: structural characterization and quantification of epoxyeicosatrienoates in plasma. Biochem Biophys Res Commun. 1992;182:1320–1325. doi: 10.1016/0006-291x(92)91877-s. [DOI] [PubMed] [Google Scholar]
  • 42.Goulitquer S, Dreano Y, Berthou F, Corcos L, Lucas D. Determination of epoxyeicosatrienoic acids in human red blood cells and plasma by GC/MS in the NICI mode. Journal of chromatography B, Analytical technologies in the biomedical and life sciences. 2008;876:83–88. doi: 10.1016/j.jchromb.2008.10.035. [DOI] [PubMed] [Google Scholar]
  • 43.Fujino H, Chen XB, Regan JW, Murayama T. Indomethacin decreases EP2 prostanoid receptor expression in colon cancer cells. Biochem Biophys Res Commun. 2007;359:568–573. doi: 10.1016/j.bbrc.2007.05.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Caughey GE, Cleland LG, Penglis PS, Gamble JR, James MJ. Roles of cyclooxygenase (COX)-1 and COX-2 in prostanoid production by human endothelial cells: selective up-regulation of prostacyclin synthesis by COX-2. Journal of immunology (Baltimore, Md : 1950) 2001;167:2831–2838. doi: 10.4049/jimmunol.167.5.2831. [DOI] [PubMed] [Google Scholar]

RESOURCES